Sigma-1 Receptor (S1R) Interaction with Cholesterol: Mechanisms of S1R Activation and Its Role in Neurodegenerative Diseases

Total Page:16

File Type:pdf, Size:1020Kb

Sigma-1 Receptor (S1R) Interaction with Cholesterol: Mechanisms of S1R Activation and Its Role in Neurodegenerative Diseases International Journal of Molecular Sciences Communication Sigma-1 Receptor (S1R) Interaction with Cholesterol: Mechanisms of S1R Activation and Its Role in Neurodegenerative Diseases Vladimir Zhemkov 1, Michal Geva 2, Michael R. Hayden 2,3 and Ilya Bezprozvanny 1,4,* 1 Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA; [email protected] 2 Prilenia Therapeutics Development LTD, Herzliya 4673304, Israel; [email protected] (M.G.); [email protected] (M.R.H.) 3 Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V6H 3V5, Canada 4 Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnic University, 195251 St. Petersburg, Russia * Correspondence: [email protected] Abstract: The sigma-1 receptor (S1R) is a 223 amino acid-long transmembrane endoplasmic retic- ulum (ER) protein. The S1R modulates the activity of multiple effector proteins, but its signaling functions are poorly understood. S1R is associated with cholesterol, and in our recent studies we demonstrated that S1R association with cholesterol induces the formation of S1R clusters. We propose that these S1R-cholesterol interactions enable the formation of cholesterol-enriched microdomains in the ER membrane. We hypothesize that a number of secreted and signaling proteins are recruited Citation: Zhemkov, V.; Geva, M.; and retained in these microdomains. This hypothesis is consistent with the results of an unbiased Hayden, M.R.; Bezprozvanny, I. Sigma-1 Receptor (S1R) Interaction screen for S1R-interacting partners, which we performed using the engineered ascorbate peroxi- with Cholesterol: Mechanisms of S1R dase 2 (APEX2) technology. We further propose that S1R agonists enable the disassembly of these Activation and Its Role in cholesterol-enriched microdomains and the release of accumulated proteins such as ion channels, Neurodegenerative Diseases. Int. J. signaling receptors, and trophic factors from the ER. This hypothesis may explain the pleotropic Mol. Sci. 2021, 22, 4082. signaling functions of the S1R, consistent with previously observed effects of S1R agonists in various https://doi.org/10.3390/ experimental systems. ijms22084082 Keywords: sigma-1 receptor; endoplasmic reticulum; mitochondria; contact sites; cholesterol; neu- Academic Editor: Carmen Abate rodegeneration; Huntington’s disease; Alzheimer’s disease; amyotrophic lateral sclerosis; drug target Received: 25 March 2021 Accepted: 13 April 2021 Published: 15 April 2021 1. Introduction Publisher’s Note: MDPI stays neutral The sigma-1 receptor (S1R) is a 223 amino acid-long transmembrane protein residing with regard to jurisdictional claims in in the endoplasmic reticulum (ER) [1–3]. S1R attracts significant attention as a potential published maps and institutional affil- drug target for treating neurological disorders [2,4–6] and cancers [6]. iations. S1R is expressed at high levels in the CNS and specifically in the cortex, basal ganglia, and motor neurons of the spinal cord and brainstem [7–10]. The S1R is a chaperone protein that is enriched at the ER/mitochondria-associated membrane (MAM), where it plays an important role in the regulation of multiple cellular mechanisms and is key to maintaining neuronal function and health. This is further supported by human genetic studies, showing Copyright: © 2021 by the authors. Licensee MDPI, Basel, Switzerland. that complete loss of function (LOF) mutations in the S1R are associated with a juvenile This article is an open access article form of amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD), while partial distributed under the terms and LOF mutations cause late onset ALS [11–14]. Thus, there is a gene dosage relationship conditions of the Creative Commons between S1R activity and the age of onset of ALS with the complete loss of S1R associated Attribution (CC BY) license (https:// with the earliest age of onset. Additional LOF mutations in the S1R cause distal hereditary creativecommons.org/licenses/by/ motor neuropathies (dHMN) [15–19]. Furthermore, S1R expression levels are reduced in 4.0/). sporadic ALS [20], Parkinson’s disease (PD), and Alzheimer’s disease (AD) patients [21,22]. Int. J. Mol. Sci. 2021, 22, 4082. https://doi.org/10.3390/ijms22084082 https://www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2021, 22, 4082 2 of 16 In preclinical models, the genetic ablation of S1R in mice exacerbated pathology and phenotypic presentation of several neurological disorders [23–25]. These results suggest that S1R plays an important role in healthy neuronal physiology. The first prototypic S1R agonist, SKF-10047, was identified in animal behavioral assays, which led to the proposed existence of sigma opioid receptors [26]. However, SKF-10047 binding to sigma binding sites was not blocked by naloxone, an opioid receptor antagonist, and displayed a different stereospecificity [27,28]. Subsequent cloning of sigma binding sites confirmed that they share no homology with opioid G protein-coupled receptors (GPCRs), as well as sharing little homology to any other mammalian protein [29,30]. Se- quence analysis revealed homology with the fungi C7–C8 sterol isomerase. While the S1R does not have isomerase activity, it contains two sterol-like binding domains as part of its ligand-binding site [29]. Recent biochemical and structural analysis indicated that the S1R is a single transmembrane domain protein with a short cytoplasmic tail and a large luminal ligand-binding domain [31,32]. It is suggested that the S1R acts as a molecu- lar chaperone, which can stabilize the native conformation of multiple client proteins in stress conditions [1,33,34]. The S1R can be activated with highly selective synthetic ligands with nanomolar affinity [34–36]. The identity of an endogenous ligand is under investiga- tion, with endogenous steroids (pregnenolone, dehydroepiandrosterone sulfate (DHEA), progesterone) being the most likely candidate [37,38], and N,N-dimethyltryptamine [39], sphingolipids [40], and more recently, choline also investigated [41]. Despite its importance in physiology and disease, the biological function of S1R is poorly understood [3]. This protein is involved in many biological processes and sig- naling pathways including maintenance of calcium homeostasis [42–45], protein fold- ing [42], stress-response [1,42,46,47], autophagy [48,49], and the regulation of cellular excitability [50–52]. The S1R modulates the activity of ion channels via protein–protein interaction [52,53]. The S1R mode of action is not coupled to any known signaling cascade and is more consistent with its role as a modulatory or adaptor protein, or, using a term first coined by Hayashi and Su, a “molecular chaperone” [34,42,54]. Several S1R-interacting partners have been identified and multiple recent reviews comprehensively summarized these S1R interactors and the S1R-induced modulation of their activities [2,3]. Apart from that, S1R is known to interact and mediate the clustering of cholesterol and ceramides in the ER, as shown in cell-based assays [55–58]. We recently demonstrated that S1R is associated with cholesterol-enriched clusters in the membranes using in vitro reconstitution approach [59]. In this review we propose a hypothesis that the biological functions of the S1R are mediated by its ability to form ER signaling cholesterol-enriched lipid microdomains, analogous to the lipid rafts in the plasma membrane [60]. 2. Intracellular Localization of the S1R S1R primarily resides in the ER membrane where it forms microdomains [42,59,61,62]. Its localization is in contrast to the uniform distribution pattern of ER markers, such as the Sec61b protein. A significant proportion of S1R is localized to MAMs, an ER sub-compartment closely associated with the mitochondria [42,59], in proximity to lipid droplets [63], and at the ER-plasma membrane (PM) junctions [59,64]. It is likely that S1R are localized to additional inter-organelle contact sites, but this has not been systematically investigated. MAMs are distinct from the rest of the ER as they contain enzymes involved in lipid synthesis, calcium signaling, cholesterol metabolism, and the ER stress-response pathways [65–69]. A detailed protein composition of MAMs was initially characterized by biochemical purifications [70,71] and more recently established using sophisticated proximity labeling approaches [72–74]. While the precise lipid composition of the MAMs has not yet been elucidated, recent evidence suggests that cholesterol and ceramide content is significantly higher in MAMs compared to the rest of the ER [57,75,76]. Therefore, MAMs can be thought of as specialized Int. J. Mol. Sci. 2021, 22, 4082 3 of 16 ER signaling domains characterized by unique protein and lipid compositions, similar to PM lipid rafts [60]. While PM lipid and protein heterogeneity was visualized using the giant plasma membrane-derived vesicle technique [77–79], only recently was a similar method to yield endomembrane-derived giant unilamellar vesicle (GUV)-like vesicles developed [80]. Using this approach, it was observed that certain, but not all ER contact sites (such as ER-mitochondria, ER-PM, and ER-lipid droplets) showed separation of the glyco- sylphosphatidylinositol (GPI) ER-targeted marker with a strong affinity for lipid-ordered phase [80]. Similar lipid and protein compartmentalization were recently observed at the inter-organelle contact sites in yeast [81], providing additional experimental
Recommended publications
  • Parkinson Disease-Associated Cognitive Impairment
    PRIMER Parkinson disease-associated cognitive impairment Dag Aarsland 1,2 ✉ , Lucia Batzu 3, Glenda M. Halliday 4, Gert J. Geurtsen 5, Clive Ballard 6, K. Ray Chaudhuri 3 and Daniel Weintraub7,8 Abstract | Parkinson disease (PD) is the second most common neurodegenerative disorder, affecting >1% of the population ≥65 years of age and with a prevalence set to double by 2030. In addition to the defining motor symptoms of PD, multiple non-motor symptoms occur; among them, cognitive impairment is common and can potentially occur at any disease stage. Cognitive decline is usually slow and insidious, but rapid in some cases. Recently, the focus has been on the early cognitive changes, where executive and visuospatial impairments are typical and can be accompanied by memory impairment, increasing the risk for early progression to dementia. Other risk factors for early progression to dementia include visual hallucinations, older age and biomarker changes such as cortical atrophy, as well as Alzheimer-type changes on functional imaging and in cerebrospinal fluid, and slowing and frequency variation on EEG. However, the mechanisms underlying cognitive decline in PD remain largely unclear. Cortical involvement of Lewy body and Alzheimer-type pathologies are key features, but multiple mechanisms are likely involved. Cholinesterase inhibition is the only high-level evidence-based treatment available, but other pharmacological and non-pharmacological strategies are being tested. Challenges include the identification of disease-modifying therapies as well as finding biomarkers to better predict cognitive decline and identify patients at high risk for early and rapid cognitive impairment. Parkinson disease (PD) is the most common movement The full spectrum of cognitive impairment occurs in disorder and the second most common neurodegenera­ individuals with PD, from subjective cognitive decline tive disorder after Alzheimer disease (AD).
    [Show full text]
  • Blarcamesine | Medchemexpress
    Inhibitors Product Data Sheet Blarcamesine • Agonists Cat. No.: HY-105296 CAS No.: 195615-83-9 Molecular Formula: C₁₉H₂₃NO • Molecular Weight: 281.39 Screening Libraries Target: Sigma Receptor; mAChR Pathway: GPCR/G Protein; Neuronal Signaling Storage: Please store the product under the recommended conditions in the Certificate of Analysis. BIOLOGICAL ACTIVITY Description Blarcamesine is an orally bioavailable Sigma-1 receptor agonist and muscarinic receptor modulator, with anticonvulsant, anti-amnesic, neuroprotective and antidepressant properties. Blarcamesine ameliorates neurologic impairments in a mouse model of Rett syndrome[1]. IC₅₀ & Target Sigma-1 receptor, muscarinic receptor[1] In Vivo Blarcamesine (10 mg/kg, 30 mg/kg; p.o.; daily; for 6.5 weeks) ameliorates motor deficits in Mecp2 HET mice with chronic administration[1]. Blarcamesine ameliorates acoustic startle deficit in Mecp2 HET mice[1]. Blarcamesine reverses deficits in visual acuity in older Mecp2 HET mice using the optokinetic response[1]. Blarcamesine produces minimal effects on body weight gain in Mecp2 HET mice[1]. MCE has not independently confirmed the accuracy of these methods. They are for reference only. Animal Model: Female Mecp2tm1.1Bird/J HET (C57 background) mice[1] Dosage: 10 mg/kg, 30 mg/kg Administration: Oral administration, daily, for 6.5 weeks Result: Reverse the deficits observed in the HET animals. REFERENCES [1]. Walter E Kaufmann, et al. ANAVEX®2-73 (Blarcamesine), a Sigma-1 Receptor Agonist, Ameliorates Neurologic Impairments in a Mouse Model of Rett Syndrome. Pharmacol Biochem Behav . 2019 Dec;187:172796. McePdfHeight Page 1 of 2 www.MedChemExpress.com Caution: Product has not been fully validated for medical applications.
    [Show full text]
  • Neural Regeneration Research
    NEURAL REGENERATION RESEARCH www.nrronline.org Additional Table 1 Agents modifying synaptic dysfunction of AD in clinical trials (ClinicalTrials.gov on November 22, 2020) Therapeutic purpose Agent ClinicalTrials.gov Mechanism of action ID/Phase/Status To improve synaptic plasticity Tacrolimus NCT04263519/2/N Calcineurin inhibitor (By enhancing LTP and decreasing Tacrolimus inhibits calcineurin-dependent LTD which may mediate synaptic loss in the AD brain. LTD) DAOI NCT03752463/2/U D-amino acid oxidase inhibitor DAOI increase the level of D-serine, a co-agonist of NMDARs. L-serine NCT03062449/2/R L-serine is the precursor of D-serine, a co-agonist of NMDARs. SAGE718 NCT04602624/2/N Positive allosteric modulator of NMDARs Bryostatin 1 NCT04538066/2/R PKC modulator AR1001 NCT03625622/2/AN PDE inhibitors BPN14770 NCT03817684/2/AN PDEs are responsible for hydrolysis of cAMP and cGMP. The inhibition of PDEs increase brain cAMP and cGMP concentrations, which activate PKA or PKG and subsequent CREB Cilostazol NCT02491268/2/AN phosphorylation in brain tissue. NCT03451591/3/R To improve synaptic plasticity AMX0035 (sodium NCT03533257/2/AN Sodium phenylbutyrate induces astrocytic BDNF and NT-3 expression via PKC-CREB pathway. phenylbutyrate and (By enhancing LTP and decreasing tauroursodeoxycholic acid LTD) combination) NEURAL REGENERATION RESEARCH www.nrronline.org Therapeutic purpose Agent ClinicalTrials.gov Mechanism of action ID/Phase/Status ANAVEX2-73 NCT03790709/3/R Sigma-1 receptor agonist (blarcamesine) NCT04314934/3/R Blarcamesine is a sigma-1 receptor agonist (high affinity), M1 receptor agonist and M2 receptor antagonist (low affinity) NCT02756858/2/AN Sigma receptor locates in endoplasmic reticulum. Agents regulate endoplasmic reticulum functions to improve synaptic plasticity by activating sigma-1 receptor.
    [Show full text]
  • Mtdls) Binding the Σ1 Receptor As Promising Therapeutics: State of the Art and Perspectives
    International Journal of Molecular Sciences Review Multi-Target Directed Ligands (MTDLs) Binding the σ1 Receptor as Promising Therapeutics: State of the Art and Perspectives Francesca Serena Abatematteo , Mauro Niso , Marialessandra Contino , Marcello Leopoldo and Carmen Abate * Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy; [email protected] (F.S.A.); [email protected] (M.N.); [email protected] (M.C.); [email protected] (M.L.) * Correspondence: [email protected]; Tel.: +39-0805442231 Abstract: The sigma-1 (σ1) receptor is a ‘pluripotent chaperone’ protein mainly expressed at the mitochondria–endoplasmic reticulum membrane interfaces where it interacts with several client proteins. This feature renders the σ1 receptor an ideal target for the development of multifunctional ligands, whose benefits are now recognized because several pathologies are multifactorial. Indeed, the current therapeutic regimens are based on the administration of different classes of drugs in order to counteract the diverse unbalanced physiological pathways associated with the pathology. Thus, the multi-targeted directed ligand (MTDL) approach, with one molecule that exerts poly- pharmacological actions, may be a winning strategy that overcomes the pharmacokinetic issues linked to the administration of diverse drugs. This review aims to point out the progress in the development of MTDLs directed toward σ1 receptors for the treatment of central nervous
    [Show full text]
  • Effects of the Sigma-1 Receptor Agonist Blarcamesine in a Murine Model Of
    www.nature.com/scientificreports OPEN Efects of the sigma‑1 receptor agonist blarcamesine in a murine model of fragile X syndrome: neurobehavioral phenotypes and receptor occupancy Samantha T. Reyes1, Robert M. J. Deacon2,3,4, Scarlett G. Guo1, Francisco J. Altimiras2,5, Jessa B. Castillo1, Berend van der Wildt1, Aimara P. Morales1, Jun Hyung Park1, Daniel Klamer6, Jarrett Rosenberg1, Lindsay M. Oberman7, Nell Rebowe6, Jefrey Sprouse6, Christopher U. Missling6, Christopher R. McCurdy8, Patricia Cogram2,3,4, Walter E. Kaufmann6,9* & Frederick T. Chin1* Fragile X syndrome (FXS), a disorder of synaptic development and function, is the most prevalent genetic form of intellectual disability and autism spectrum disorder. FXS mouse models display clinically‑relevant phenotypes, such as increased anxiety and hyperactivity. Despite their availability, so far advances in drug development have not yielded new treatments. Therefore, testing novel drugs that can ameliorate FXS’ cognitive and behavioral impairments is imperative. ANAVEX2‑73 (blarcamesine) is a sigma‑1 receptor (S1R) agonist with a strong safety record and preliminary efcacy evidence in patients with Alzheimer’s disease and Rett syndrome, other synaptic neurodegenerative and neurodevelopmental disorders. S1R’s role in calcium homeostasis and mitochondrial function, cellular functions related to synaptic function, makes blarcamesine a potential drug candidate for FXS. Administration of blarcamesine in 2‑month‑old FXS and wild type mice for 2 weeks led to normalization in two key neurobehavioral phenotypes: open feld test (hyperactivity) and contextual fear conditioning (associative learning). Furthermore, there was improvement in marble‑burying (anxiety, perseverative behavior). It also restored levels of BDNF, a converging point of many synaptic regulators, in the hippocampus.
    [Show full text]
  • A Business Development Look at the Cns Landscape
    PULLAN’S PIECES, PART II A BUSINESS DEVELOPMENT LOOK AT THE CNS LANDSCAPE By Linda M. Pullan, PhD, Pullan Consulting The unmet needs in CNS are huge. With one hundred million Americans affected by neurological disorders and 43 million Americans suffering from some type of mental illness, the societal impact of CNS afflictions is astronomical. There may be no other therapeutic area where the unmet needs are bigger. What’s more, these diseases are often accompanied by a “loss of self,” which in itself is devastating. 100M 43.4M Americans American affected by adults have neurologica&l a mental disorders illness (MID Report 2018) (MID Report 2017) ShareVault and Pullan Consulting The numbers of indications with drugs in development are also impressive, both in the categories of neurology and psychiatry. Indications shown in orange have over 7 million patients. Chronic Orphan Neurology • ADHD y y Neurological • Alcohol • ALS r addiction g • Alzheimer’s • Ataxias t • Anorexia • Chronic • Batten Disease a o Nervosa i l Inflammatory • Broca Aphasia • Autism Demyelinating • Canavan Disease h o • Binge eating Polyneuropathy • Carnosinemia r c • Bipolar • Complex • Cerebral Palsy Disorder u Regional Pain • Dravet y • Dementia Syndrome Syndrome s e • Depression • Dyskinesia • Friedreich Ataxia P • Insomnia • Epilepsy • Globoid Cell N • Narcolepsy • Fibromyalgia Leukodystrophy • Panic Disorder • HD • Syringomyelia • PTSD • MCI • Tardive • Schizophrenia • Migraine Dyskinesia • Tourette • MS • Hyperkalemic Syndrome • Multi-focal Motor Periodic Neuropathy
    [Show full text]
  • Bi-Phasic Dose Response in the Preclinical and Clinical Developments of Sigma-1 Receptor Ligands for the Treatment of Neurodegenerative Disorders Tangui Maurice
    Bi-phasic dose response in the preclinical and clinical developments of sigma-1 receptor ligands for the treatment of neurodegenerative disorders Tangui Maurice To cite this version: Tangui Maurice. Bi-phasic dose response in the preclinical and clinical developments of sigma-1 receptor ligands for the treatment of neurodegenerative disorders. Expert Opinion on Drug Discovery, Informa Healthcare, 2020, 10.1080/17460441.2021.1838483. hal-03020731 HAL Id: hal-03020731 https://hal.archives-ouvertes.fr/hal-03020731 Submitted on 24 Nov 2020 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Bi-phasic dose response in the preclinical and clinical developments of sigma-1 receptor ligands for the treatment of neurodegenerative disorders Tangui MAURICE MMDN, Univ Montpellier, EPHE, INSERM, UMR_S1198, Montpellier, France Correspondance: Dr T. Maurice, MMDN, INSERM UMR_S1198, Université de Montpellier, CC105, place EuGène Bataillon, 34095 Montpellier cedex 5, France. Tel.: +33/0 4 67 14 32 91. E-mail: [email protected] Abstract Introduction: The sigma-1 receptor (S1R) is attracting much attention as a target for disease- modifying therapies in neurodegenerative diseases. It is a highly conserved protein, present in plasma and endoplasmic reticulum (ER) membranes and enriched in mitochondria-associated ER membranes (MAMs).
    [Show full text]
  • Horizon Scanning Status Report December 2020
    PCORI Health Care Horizon Scanning System Volume 2 Issue 4 Horizon Scanning Status Report December 2020 Prepared for: Patient-Centered Outcomes Research Institute 1828 L St., NW, Suite 900 Washington, DC 20036 Contract No. MSA-HORIZSCAN-ECRI-ENG-2018.7.12 Prepared by: ECRI Institute 5200 Butler Pike Plymouth Meeting, PA 19462 Investigators: Randy Hulshizer, MA, MS Damian Carlson, MS Christian Cuevas, PhD Andrea Druga, MSPAS, PA-C Marcus Lynch, PhD, MBA Misha Mehta, MS Prital Patel, MPH Brian Wilkinson, MA Donna Beales, MLIS Jennifer De Lurio, MS Eloise DeHaan, BS Eileen Erinoff, MSLIS Cassia Hulshizer, BBA Madison Kimball, MS Maria Middleton, MPH Melinda Rossi, BS Diane Robertson, BA Kelley Tipton, MPH Rosemary Walker, MLIS Andrew Furman, MD, MMM, FACEP Statement of Funding and Purpose This report incorporates data collected during implementation of the Patient-Centered Outcomes Research Institute (PCORI) Health Care Horizon Scanning System, operated by ECRI under contract to PCORI, Washington, DC (Contract No. MSA-HORIZSCAN-ECRI-ENG-2018.7.12). The findings and conclusions in this document are those of the authors, who are responsible for its content. No statement in this report should be construed as an official position of PCORI. An intervention that potentially meets inclusion criteria might not appear in this report simply because the Horizon Scanning System has not yet detected it or it does not yet meet the inclusion criteria outlined in the PCORI Health Care Horizon Scanning System: Horizon Scanning Protocol and Operations Manual. Inclusion or absence of interventions in the horizon scanning reports will change over time as new information is collected; therefore, inclusion or absence should not be construed as either an endorsement or a rejection of specific interventions.
    [Show full text]
  • CTAD POSTER 2019-1.Pdf
    The Journal of Prevention of Alzheimer’s Disease - JPAD© © Serdi and Springer Nature Switzerland AG 2019 Volume 6, Supplement 1, 2019 and memory. Third, we determined associations between demographic variables (gender, race, ethnicity, and education) POSTERS and registry participation (withdrawal and task completion) using logistic regression methods. Results: Of the 17,073 BHR Theme: CLINICAL TRIALS METHODOLOGY participants age 65+, 65% were female, 85% identified as Caucasian, 91% identified as non-Hispanic/Latino, and 68% had P001- UNDERREPRESENTED ELDERS IN THE BRAIN a bachelor’s degree or higher. Compared to the Census data, HEALTH REGISTRY: US REPRESENTATIVENESS the BHR underrepresented males (∆=-9.39%, p<.0001, V=0.003) AND REGISTRY BEHAVIOR. M.T. Ashford1,2,*, and Hispanic/Latino participants (∆=-5.81%, p<.0001, V=0.005). J. Eichenbaum1,2,3, T. Williams1,2,3, J. Fockler2,3, Non-Caucasians were also underrepresented (p<.0001, V=0.005), M. Camacho1,2, A. Ulbricht2,3, D. Flenniken1,2, specifically Black/African American participants (∆=-6.06%), D. Truran1,2, R.S. Mackin2,4, M.W. Weiner1,2,3, R.L. Nosheny2,4 Asian participants (∆=-2.35%), Native American and Alaska ((1) Northern California Institute for Research and Education Native participants (-0.31%), and participants from some other (NCIRE) - San Francisco (United States); (2) Department race (∆=-0.26%). Participants with lower education levels were of Veterans Affairs Medical Center, Center for Imaging also underrepresented (p<.0001, V=0.02), including those with and Neurodegenerative Diseases - San Francisco (United a community college degree or associate’s degree (∆=-1.37%), States); (3) Department of Radiology and Biomedical a high school degree (∆=-25.27%), or lower (∆=-15.26%).
    [Show full text]
  • Horizon Scanning Status Report June 2020 Prepared For: Patient-Centered Outcomes Research Institute 1828 L St., NW, Suite 900 Washington, DC 20036
    PCORI Health Care Horizon Scanning System Volume 2 Issue 2 Horizon Scanning Status Report June 2020 Prepared for: Patient-Centered Outcomes Research Institute 1828 L St., NW, Suite 900 Washington, DC 20036 Contract No. MSA-HORIZSCAN-ECRI-ENG-2018.7.12 Prepared by: ECRI Institute 5200 Butler Pike Plymouth Meeting, PA 19462 Investigators: Randy Hulshizer, MA, MS Damian Carlson, MS Christian Cuevas, PhD Andrea Druga, PA-C Marcus Lynch, PhD Misha Mehta, MS Brian Wilkinson, MA Donna Beales, MLIS Jennifer De Lurio, MS Eloise DeHaan, BS Eileen Erinoff, MSLIS Madison Kimball, MS Maria Middleton, MPH Diane Robertson, BA Kelley Tipton, MPH Rosemary Walker, MLIS Karen Schoelles, MD, SM Statement of Funding and Purpose This report incorporates data collected during implementation of the Patient-Centered Outcomes Research Institute (PCORI) Health Care Horizon Scanning System, operated by ECRI Institute under contract to PCORI, Washington, DC (Contract No. MSA-HORIZSCAN-ECRI-ENG- 2018.7.12). The findings and conclusions in this document are those of the authors, who are responsible for its content. No statement in this report should be construed as an official position of PCORI. An intervention that potentially meets inclusion criteria might not appear in this report simply because the Horizon Scanning System has not yet detected it or it does not yet meet inclusion criteria outlined in the PCORI Health Care Horizon Scanning System: Horizon Scanning Protocol and Operations Manual. Inclusion or absence of interventions in the horizon scanning reports will change over time as new information is collected; therefore, inclusion or absence should not be construed as either an endorsement or rejection of specific interventions.
    [Show full text]
  • Lung Defects Contribute to Respiratory Symptoms in a Mecp2-Mutant Mouse Model of Rett Syndrome
    Lung Defects Contribute to Respiratory Symptoms in a Mecp2-Mutant Mouse Model of Rett Syndrome by Neeti Vashi A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Molecular Genetics University of Toronto © Copyright by Neeti Vashi 2021 Lung Defects Contribute to Respiratory Symptoms in a Mecp2- Mutant Mouse Model of Rett Syndrome Neeti Vashi Doctor of Philosophy Molecular Genetics University of Toronto 2021 Abstract Rett syndrome (RTT) is a progressive neuro-metabolic disorder caused by mutations in the X- linked gene, methyl-CpG-binding protein 2 (MECP2). After a period of seemingly normal post- natal development, RTT patients experience a developmental regression, consisting of loss of acquired verbal and motor skills, stereotypic hand movements, respiratory abnormalities, and seizures. Respiratory impairment causes up to 80% of premature patient death; despite this, lung pathology in RTT is understudied and respiratory symptoms are currently attributed to neuronal loss of MECP2. To study the Mecp2-deficient lung, we utilized a Mecp2-mutant mouse model that recapitulates many features of RTT. I found striking lipid metabolism abnormalities in the lungs of Mecp2-mutant mice, including increased cholesterol and triglycerides and decreased phosphatidylcholines. My single cell RNA-sequencing and chromatin immunoprecipitation experiments showed that lipogenesis is increased due to decreased binding of the nuclear repressor coreceptor 1/2 (NCOR1/2) complex in the promoters of its target genes in the absence of MECP2, leading to their upregulation. I also showed that lung AE2 cell-specific depletion of Mecp2 is sufficient to cause lung lipid metabolism abnormalities and respiratory symptoms. In contrast, hindbrain neuron-specific deletion of Mecp2, which removes Mecp2 from the neuronal respiratory control center, imparted a different respiratory phenotype.
    [Show full text]
  • An Emerging Role for Sigma-1 Receptors in the Treatment of Developmental and Epileptic Encephalopathies
    International Journal of Molecular Sciences Review An Emerging Role for Sigma-1 Receptors in the Treatment of Developmental and Epileptic Encephalopathies Parthena Martin 1, Thadd Reeder 1, Jo Sourbron 2 , Peter A. M. de Witte 3 , Arnold R. Gammaitoni 1 and Bradley S. Galer 1,* 1 Zogenix, Inc., Emeryville, CA 94608, USA; [email protected] (P.M.); [email protected] (T.R.); [email protected] (A.R.G.) 2 University Hospital KU Leuven, 3000 Leuven, Belgium; [email protected] 3 Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences at KU Leuven, 3000 Leuven, Belgium; [email protected] * Correspondence: [email protected]; Tel.: +1-484-675-5884 Abstract: Developmental and epileptic encephalopathies (DEEs) are complex conditions character- ized primarily by seizures associated with neurodevelopmental and motor deficits. Recent evidence supports sigma-1 receptor modulation in both neuroprotection and antiseizure activity, suggesting that sigma-1 receptors may play a role in the pathogenesis of DEEs, and that targeting this receptor has the potential to positively impact both seizures and non-seizure outcomes in these disorders. Recent studies have demonstrated that the antiseizure medication fenfluramine, a serotonin-releasing drug that also acts as a positive modulator of sigma-1 receptors, reduces seizures and improves everyday executive functions (behavior, emotions, cognition) in patients with Dravet syndrome and Lennox-Gastaut syndrome. Here, we review the evidence for sigma-1 activity in reducing Citation: Martin, P.; Reeder, T.; Sourbron, J.; de Witte, P.A.M.; seizure frequency and promoting neuroprotection in the context of DEE pathophysiology and clinical Gammaitoni, A.R.; Galer, B.S.
    [Show full text]